Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 14, 2024; 30(34): 3850-3855
Published online Sep 14, 2024. doi: 10.3748/wjg.v30.i34.3850
Sirtuin 1 in regulating the p53/glutathione peroxidase 4/gasdermin D axis in acute liver failure
Swati Katoch, Vikram Patial, Division of Dietetics and Nutrition Technology, Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India
Swati Katoch, Vikram Patial, Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
ORCID number: Swati Katoch (0000-0001-9120-6327); Vikram Patial (0000-0002-4912-9871).
Author contributions: Katoch S collected the literature and wrote the manuscript; Patial V contributed to designing, manuscript writing and editing; Both authors have read and approved the final manuscript.
Supported by CSIR, India, No. MLP0204.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Vikram Patial, PhD, Principal Scientist, Associate Professor, Division of Dietetics and Nutrition Technology, Institute of Himalayan Bioresource Technology, Post Box No. 6, Palampur 176061, Himachal Pradesh, India. vikrampatial@ihbt.res.in
Received: March 15, 2024
Revised: August 14, 2024
Accepted: August 21, 2024
Published online: September 14, 2024
Processing time: 178 Days and 19.7 Hours

Abstract

In this editorial, we comment on the article by Zhou et al. The study reveals the connection between ferroptosis and pyroptosis and the effect of silent information regulator sirtuin 1 (SIRT1) activation in acute liver failure (ALF). ALF is characterized by a sudden and severe liver injury resulting in significant hepatocyte damage, often posing a high risk of mortality. The predominant form of hepatic cell death in ALF involves apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Glutathione peroxidase 4 (GPX4) inhibition sensitizes the cell to ferroptosis and triggers cell death, while Gasdermin D (GSDMD) is a mediator of pyroptosis. The study showed that ferroptosis and pyroptosis in ALF are regulated by blocking the p53/GPX4/GSDMD pathway, bridging the gap between the two processes. The inhibition of p53 elevates the levels of GPX4, reducing the levels of inflammatory and liver injury markers, ferroptotic events, and GSDMD-N protein levels. Reduced p53 expression and increased GPX4 on deletion of GSDMD indicated ferroptosis and pyroptosis interaction. SIRT1 is a NAD-dependent deacetylase, and its activation attenuates liver injury and inflammation, accompanied by reduced ferroptosis and pyroptosis-related proteins in ALF. SIRT1 activation also inhibits the p53/GPX4/GSDMD axis by inducing p53 acetylation, attenuating LPS/D-GalN-induced ALF.

Key Words: Acute liver failure; Ferroptosis; Gasdermin D; Pyroptosis; p53; Silent information regulator sirtuin 1

Core Tip: Ferroptosis and pyroptosis are crucial modes of hepatic cell death in acute liver failure (ALF), and their close association leads to the sudden progression of liver failure. Silent information regulator sirtuin 1 inhibits p53 activity by deacetylation, thereby promoting cell survival. Inhibition of p53 activity affects downstream regulators like Glutathione peroxidase 4 (GPX4) and Gasdermin D (GSDMD). The overexpression of GPX4 and reduced levels of GSDMD protect the cell from pyroptosis and ferroptosis, indicating the close link between these mechanisms in ALF.



INTRODUCTION

Acute liver failure (ALF), also referred to as fulminant hepatic failure, is a rare yet serious condition characterized by the rapid deterioration of liver function, often occurring within a span of days to weeks. It can be caused by various factors, including drug overdose, viral infections, and autoimmune diseases[1]. This rapid deterioration of liver function can lead to severe complications, including hepatic encephalopathy, coagulopathy (bleeding disorders), and multi-organ failure[2]. According to a report, patients with ALF had 40.02% mortality[3]. ALF accounts for 6% of all liver disease-related deaths in the United States, and 3000 cases are reported annually. It is more common in Americans than other racial groups and more common in women than men. The peak incidence of ALF is at a relatively young age of 35 years in women and 45 years in men[4]. Drug-induced liver injury is the most common cause of ALF in developed countries, accounting for over 50% of cases. It can be caused by various medications, including acetaminophen (paracetamol) overdose, certain antibiotics, anti-epileptic drugs, and herbal remedies[5]. Recent studies have proposed several cell death pathways, including ferroptosis, pyroptosis, necroptosis, and apoptosis, that are important in the development of ALF[6,7]. Among them, the two distinguishing programmed cell death processes that are distinct from autophagy, apoptosis, and necrosis are ferroptosis and pyroptosis. Both of these processes are vital for the development of ALF as they each modulate distinct inflammatory and immunological responses[8].

The main mechanism behind ferroptosis is reactive oxygen species (ROS)-dependent regulated cell death, with lipid peroxidation, intracellular iron overload, and decreased glutathione peroxidase 4 (GPX4) activity serving as key indicators. The Fenton process results in the production of ROS, which changes hydrogen peroxide into hydroxyl radicals and subsequently causes lipid peroxidation[9]. ROS growth is regulated by the inhibitory activity of GPX4, an enzyme that uses hydrogen ions to neutralize lipid peroxides and hydrogen peroxide. Reduced glutathione (GSH) is transformed into glutathione disulfide (GSSG) by GPX4[10]. In addition to GPX4’s basic function of preserving ROS levels, numerous other pathways are necessary to sustain GPX’s antioxidant activity. System Xc- is a heterodimeric antiport system comprising the subunits SLC3A2 and SLC7A11 that import cystine and output glutamate. The imported cystine is converted into cysteine and GSH to maintain redox equilibrium and shield the cells from ferroptosis[11]. The primary mechanism that sustains the effectiveness of GPX antioxidant action is System Xc-, which inhibits a sequence of processes that lead to a decrease in GSH levels, lipid peroxidation, and, ultimately, ferroptosis[12].

Due to p53’s transcriptional inhibition of SLC7A11, cells are more susceptible to ferroptosis and absorb cystine less efficiently. The cellular response to various triggers of stress, such as hypoxia, starvation, DNA damage, and oncogene activation, is greatly influenced by p53. Furthermore, by inhibiting cystine metabolism and ROS activity, p53 promotes ferroptosis[13].

Pyroptosis is an acute inflammatory form of programmed cell death that is mostly provoked by either canonical or noncanonical inflammasomes. It is characterized by the morphological enlargement of cells followed by lysis, ultimately releasing intracellular material[14]. The liver is constantly exposed to various gut-derived microbial particles known as pathogen-associated molecular patterns (PAMPs) due to the tight connection between portal circulation and the intestines. PAMPs excite local immune cells[15]. When risk signals are detected, intracellular multiprotein complexes known as canonical inflammasomes are formed. These inflammasomes activate caspase-1, which leads to the maturation of interleukin-1 beta (IL-1β), IL-18, and IL-37, ultimately causing pyroptosis. Caspase-11 is activated by noncanonical inflammasomes, causing pyroptosis[16]. Gasdermin D (GSDMD) is a pore-forming protein that incites pro-inflammatory cytokine release and causes pyroptosis[17]. PAMPs stimulate distinct inflammasomes to initiate caspase-1, which subsequently cleaves GSDMD, the pyroptosis executor, into its active N terminal and inactive C terminal. The C-terminal regulatory domain (GSDMD-C) and the pyroptotic N-terminal domain (GSDMD-N) of GSDMD are joined by a linker domain. Inflammatory caspases (caspase-1, caspase-4, and caspase-5) break the linker domain of GSDMD, releasing GSDMD-N from its autoinhibitory GSDMD-C. GSDMD-N demonstrates a remarkable resemblance to membrane lipids, where it oligomerizes to produce pores with a diameter of around 20 nm, causing swelling and cell lysis, while GSDMD-C stays in the cytoplasm[18].

Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, is believed to protect cells from oxidative stress injury by mediating Nrf2 production and its downstream targets[19]. SIRT1 plays a complex function in stress responses, energy metabolism, inflammation, and redox balance through the acetylation and deacetylation of certain transcription factors and proteins, including p53[20]. Zhou et al[21] discussed the method by which SIRT1 reduces ALF and how it is linked to hepatocyte death that occurs widely and involves ferroptosis and pyroptosis[21].

ROLE OF p53/GPX4/GSDMD AXIS IN ACUTE LIVER FAILURE

Zhou et al[21] have carefully examined the process by which p53/GPX4/GSDMD signaling pathway blockade caused by SIRT1 activation decreases ferroptosis and pyroptosis in ALF (Figure 1). The increased activity of the enzyme biomarkers alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in human ALF is linked to elevated levels of inflammatory factors, such as tumor necrosis factor-α (TNFα), IL-1β, and IL-6. Furthermore, ferroptosis-related antioxidant protein levels of GPX4 and SLC7A11 drop in ALF, worsening iron deposition and leading to increased expression of GSDMD[21]. It is unclear how ferroptosis and pyroptosis are related to fulminant hepatitis in humans, despite the fact that the etiology of LPS/D-GalN-induced ALF in mice is quite similar to that in humans.

Figure 1
Figure 1 The activation of sirtuin 1 suppresses ferroptosis and pyroptosis in acute liver failure by inhibiting p53, glutathione peroxidase 4, and gasdermin D. The silent information regulator sirtuin 1 blocks p53 deacetylation and inhibits the glutathione peroxidase 4 (GPX4)/gasdermin D (GSDMD) signaling pathway, which in turn reduces hepatic iron overload and the inflammatory response. Ferroptosis and pyroptosis in acute liver failure are reduced by blocking the p53/GPX4/GSDMD signaling pathway. SIRT1: Silent information regulator sirtuin 1; SLC7A11: Solute carrier family 7a member 11; SLC3A2: Solute carrier family 3a member 2; GSH: Glutathione; GPX4: Glutathione peroxidase 4; GSDMD: Gasdermin D; IL1β: Interleukin-1 beta; ROS: Reactive oxygen species.

GSDMD and GPX4 are essential for ferroptosis and pyroptosis to occur. Conditions leading to GPX4 instability or suppression make cells more susceptible to ferroptosis or even lead to ferroptotic cell death, whereas sufficient GPX4 activity and GSH synthesis are necessary to prevent ferroptosis[22]. A recent study in a murine model found that ferroptosis was significantly involved in liver failure produced by acetaminophen. The results showed higher levels of TNFα, interleukins, and PTGS2, a well-known marker for ferroptosis. This process was considerably suppressed by ferrostatin-1, an inhibitor that is unique to ferroptosis. Ferroptosis may be a viable therapeutic target for liver failure, according to these findings[23]. Increased ROS levels, decreased System Xc-activity, and suppressed GPX4 levels are the main inducers of ferroptosis[24]. The LPS/D-GalN-induced liver failure model was reported to have decreased levels of SLC7A11, HO-1, and GPX4, along with increased ROS production, indicating System Xc- involvement in ferroptosis[25]. The inflammatory action is further prolonged by GSDMD-mediated hepatocyte pyroptosis, which stimulates macrophages by escalating the levels of monocyte chemotactic protein 1/CC chemokine receptor-2[18]. A study reported that pre-administration of oyster-derived Tyr-Ala (YA) peptide improves the elevated levels of GSDMD, along with its regulatory factors like caspase-1, IL-1β and TNFα in the LPS/D-GalN induced ALF model[26]. Many researchers reported that GSDMD gene knockout ameliorates pyroptosis and ferroptosis with reduced inflammatory reactions and hepatocyte loss, which improves ALF[27,28].

The non-classical process of pyroptotic cell death is mediated by caspase-11, which is implicated in myeloid cell knockout[29]. This process was seen in mice lacking the GPX4 gene that were susceptible to a deadly infection and had septic myeloid cells. GPX4 suppressed the caspase-1-dependent NLRP3 inflammasome, suggesting a comprehensive role of GPX4 against pyroptosis[30]. According to a different study, 3,4-dihydroxyphenylethyl alcohol glycoside (DAG) prevents hepatocyte ferroptosis and pyroptosis, which lowers ALF in mice caused by acetaminophen. DAG further decreased the levels of proinflammatory cytokines, histological changes, hepatic neutrophil infiltration, and serum ALT and AST. It also suppressed the levels of MDA adducts and the depletion of GSH, CAT, and SOD enzymes. In vitro, in mouse AML12 hepatocytes exposed to acetaminophen, DAG demonstrated a dose-dependent suppression of proinflammatory factors (IL-1β and IL18), ROS, and the reduction of GSH depletion. Interestingly, in liver tissues and AML12 hepatocytes, DAG increased the expression of GPX4 and decreased that of HO-1, NLRP3, caspase-1, and GSDMD[31].

The p53 tumor suppressor protein is a critical cell cycle and apoptosis regulator. p53 regulates the progression of liver injury and regeneration after acetaminophen overdose[32]. p53 knockout mice (p53KO) showed prolonged steatosis, lower mitochondrial DNA content, and reduced expression of mitochondrial transcription factor A and mitochondrial complexes in acetaminophen-induced liver injury. It also altered metabolic homeostasis and activated proinflammatory and proliferative signaling. Prolonged steatosis in the p53KO group was also linked with p53 targets related to fatty acid balance, SREBP2 protein, and GAMT mRNA expression[32]. Zhou et al’s study[21] demonstrated that the inhibition of p53 and increased GPX4 in the ALF mouse model reduced the inflammatory responses, AST and ALT levels, and ferroptotic events (depletion of GPX4, GSH, and SLC7A11, as well as iron buildup). A cross-talk between ferroptosis and pyroptosis was observed, as evidenced by the reduction of p53 expression and the elevation of GPX4 following GSDMD knockout. Furthermore, ALT and AST levels, ferroptosis markers, and GSDMD were markedly enhanced in response to GPX4 knockdown[21]. The findings suggest the possibility of a positive feedback loop and the alleviation of ferroptosis and pyroptosis in ALF caused by disrupting the p53/GPX4/GSDMD signaling pathway. Another study showed that altering GPX4 protein expression did not influence p53 levels; however, it did operate indirectly by controlling GSDMD, indicating that GPX4 is a downstream regulator of p53. p53-driven ferroptosis is produced in a GPX4-independent manner, and p53 levels remain unaffected by the deletion of ACSL4 and GPX4. However, p53 transcription decreases with GPX4 augmentation, indicating the correlation between GPX4, GSDMD, and p53 in ferroptosis and pyroptosis[33].

SIRT1, a NAD-dependent deacetylase, mediates the function of p53 by direct deacetylation. According to one study, negative regulation of SIRT1 exacerbated the acute hepatic proinflammatory response and induced pyroptosis[34]. However, SIRT1 overexpression eliminated p53 acetylation levels and decreased the release of hepatic enzymes, hepatic oxidative stress, and inflammation in acetaminophen-induced liver injury[35]. The immediate target gene of p53 is microRNA-34a (miR-34a), which concurrently activates p53 via SIRT1. The miR-34a/SIRT1/p53 signaling pathway, crucial for cell division and death, creates a positive feedback loop in which p53 stimulates miR-34a, and miR-34a promotes p53 by blocking SIRT1. Human patients or animal models with several liver disorders, including liver fibrosis, have been reported with higher expression levels of miR-34a[36]. SIRT1 overexpression attenuates liver fibrosis by decreasing p53 acetylation and caspase activation in apoptosis[37]. In the case of myocardial ischemia-reperfusion injury, SIRT1 activation was found to reduce ferroptosis-induced cardiac cell death by overexpression of SLC7A11 and inhibition of p53, indicating the close link of the SIRT1/p53/SLC7A11 axis[38]. Mice treated with resveratrol, a small-molecule SIRT1 activator, did not suffer hepatic ischemia-reperfusion injury[39]. Overall, all the findings establish a cross-talk between ferroptosis and pyroptosis with their primary upstream and downstream regulatory mechanisms. The activation of SIRT1 inhibits p53 and GSDMD activity while stimulating GPX4 action, overall blocking the p53/GPX4/GSDMD axis and protecting the cell from ferroptosis and pyroptosis.

CLINICAL IMPLICATIONS OF ACUTE LIVER FAILURE

ALF can have significant clinical implications, ranging from mild symptoms to life-threatening complications. Some common implications related to ALF are hepatic dysfunction, coagulopathy, multi-organ dysfunction, hepatic encephalopathy, infection susceptibility, and various long-term complications[2]. Early recognition, prompt diagnosis, and appropriate management are essential to improve outcomes and reduce morbidity and mortality associated with ALF[3]. Certain naturally occurring substances, such as resveratrol in red wine, have been recognized as SIRT1 agonists. Resveratrol has drawn interest because of its potential health benefits and capacity to activate SIRT1 and replicate the effects of caloric restriction, which have been linked to longer lifespans in various organisms[39]. Synthetic SIRT1 agonist development has also been the focus of researchers for possible medicinal uses. Compared to natural substances, these synthesized molecules may provide a more robust and tailored activation of SIRT1. Researchers are looking at the potential of SIRT1 agonists in cancer, neurological diseases, and metabolic disorders, among other areas. The clinical advantages and safety of SIRT1 agonists in humans are still being assessed through ongoing research and clinical trials, even though SIRT1 activation shows promising results in preclinical studies. It is important to remember that SIRT1 is a multifaceted molecule whose function in human health is currently being studied. This work demonstrated a connection between the upstream regulatory processes and ferroptosis and pyroptosis. More investigations are required to ascertain the possible therapeutic advantages of targeting SIRT1 to treat various metabolic disorders.

CONCLUSION

To sum up, this editorial examines the data supporting the notion that ferroptosis and pyroptosis are essential hepatocyte death mechanisms in ALF, and that the interplay between these cell death mechanisms promotes the development of ALF. The findings suggest that SIRT1 and p53 can regulate each other in a feedback loop. While SIRT1 has been shown to deacetylate and inhibit p53 activity, thereby promoting cell survival and inhibiting apoptosis, p53 activation can lead to increased expression of SIRT1, which may have various downstream effects on cellular processes. Considering the reported shreds of evidence, we can attenuate ALF by blocking the p53/GPX4/GSDMD signaling pathway and activating SIRT1, which results in reduced ferroptosis and pyroptosis. This signaling pathway, especially SIRT1, might be a promising therapeutic target for liver failure.

ACKNOWLEDGEMENTS

The authors are thankful to the Director, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India, for his continuous support (Communication no. 5627).

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Shahid N S-Editor: Li L L-Editor: Webster JR P-Editor: Yuan YY

References
1.  Stravitz RT, Lee WM. Acute liver failure. Lancet. 2019;394:869-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 435]  [Article Influence: 87.0]  [Reference Citation Analysis (0)]
2.  Bernal W, Jalan R, Quaglia A, Simpson K, Wendon J, Burroughs A. Acute-on-chronic liver failure. Lancet. 2015;386:1576-1587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 216]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
3.  Hernaez R, Kramer JR, Liu Y, Tansel A, Natarajan Y, Hussain KB, Ginès P, Solà E, Moreau R, Gerbes A, El-Serag HB, Kanwal F. Prevalence and short-term mortality of acute-on-chronic liver failure: A national cohort study from the USA. J Hepatol. 2019;70:639-647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (1)]
4.  Singh T, Gupta N, Alkhouri N, Carey WD, Hanouneh IA. A guide to managing acute liver failure. Cleve Clin J Med. 2016;83:453-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
5.  Lee WM. Drug-induced acute liver failure. Clin Liver Dis. 2013;17:575-586, viii.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 162]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
6.  Xie D, Ouyang S. The role and mechanisms of macrophage polarization and hepatocyte pyroptosis in acute liver failure. Front Immunol. 2023;14:1279264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  Mao L, Zhao T, Song Y, Lin L, Fan X, Cui B, Feng H, Wang X, Yu Q, Zhang J, Jiang K, Wang B, Sun C. The emerging role of ferroptosis in non-cancer liver diseases: hype or increasing hope? Cell Death Dis. 2020;11:518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 77]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
8.  Stockwell BR, Jiang X, Gu W. Emerging Mechanisms and Disease Relevance of Ferroptosis. Trends Cell Biol. 2020;30:478-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 625]  [Article Influence: 156.3]  [Reference Citation Analysis (0)]
9.  Münzel T, Gori T, Keaney JF Jr, Maack C, Daiber A. Pathophysiological role of oxidative stress in systolic and diastolic heart failure and its therapeutic implications. Eur Heart J. 2015;36:2555-2564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 255]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
10.  Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B 3rd, Stockwell BR. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060-1072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4711]  [Cited by in F6Publishing: 9050]  [Article Influence: 754.2]  [Reference Citation Analysis (1)]
11.  Liu MR, Zhu WT, Pei DS. System Xc(-): a key regulatory target of ferroptosis in cancer. Invest New Drugs. 2021;39:1123-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 103]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
12.  Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kalivas PW, Massie A, Smolders I, Methner A, Pergande M, Smith SB, Ganapathy V, Maher P. The cystine/glutamate antiporter system x(c)(-) in health and disease: from molecular mechanisms to novel therapeutic opportunities. Antioxid Redox Signal. 2013;18:522-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 533]  [Cited by in F6Publishing: 656]  [Article Influence: 59.6]  [Reference Citation Analysis (0)]
13.  Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R, Gu W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1173]  [Cited by in F6Publishing: 1967]  [Article Influence: 218.6]  [Reference Citation Analysis (0)]
14.  Fang Y, Tian S, Pan Y, Li W, Wang Q, Tang Y, Yu T, Wu X, Shi Y, Ma P, Shu Y. Pyroptosis: A new frontier in cancer. Biomed Pharmacother. 2020;121:109595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 571]  [Article Influence: 114.2]  [Reference Citation Analysis (0)]
15.  Wang J, Dong R, Zheng S. Roles of the inflammasome in the gutliver axis (Review). Mol Med Rep. 2019;19:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (1)]
16.  Yuan YY, Xie KX, Wang SL, Yuan LW. Inflammatory caspase-related pyroptosis: mechanism, regulation and therapeutic potential for inflammatory bowel disease. Gastroenterol Rep (Oxf). 2018;6:167-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 78]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
17.  Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526:660-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2573]  [Cited by in F6Publishing: 3879]  [Article Influence: 431.0]  [Reference Citation Analysis (0)]
18.  Li H, Zhao XK, Cheng YJ, Zhang Q, Wu J, Lu S, Zhang W, Liu Y, Zhou MY, Wang Y, Yang J, Cheng ML. Gasdermin D-mediated hepatocyte pyroptosis expands inflammatory responses that aggravate acute liver failure by upregulating monocyte chemotactic protein 1/CC chemokine receptor-2 to recruit macrophages. World J Gastroenterol. 2019;25:6527-6540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (1)]
19.  Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid Redox Signal. 2018;28:643-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 351]  [Cited by in F6Publishing: 482]  [Article Influence: 80.3]  [Reference Citation Analysis (0)]
20.  Liu X, Yang X, Han L, Ye F, Liu M, Fan W, Zhang K, Kong Y, Zhang J, Shi L, Chen Y, Zhang X, Lin S. Pterostilbene alleviates polymicrobial sepsis-induced liver injury: Possible role of SIRT1 signaling. Int Immunopharmacol. 2017;49:50-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 32]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
21.  Zhou XN, Zhang Q, Peng H, Qin YJ, Liu YH, Wang L, Cheng ML, Luo XH, Li H. Silent information regulator sirtuin 1 ameliorates acute liver failure via the p53/glutathione peroxidase 4/gasdermin D axis. World J Gastroenterol. 2024;30:1588-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Wang Y, Zhang H, Chen Q, Jiao F, Shi C, Pei M, Lv J, Zhang H, Wang L, Gong Z. TNF-α/HMGB1 inflammation signalling pathway regulates pyroptosis during liver failure and acute kidney injury. Cell Prolif. 2020;53:e12829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 113]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
23.  Yamada N, Karasawa T, Kimura H, Watanabe S, Komada T, Kamata R, Sampilvanjil A, Ito J, Nakagawa K, Kuwata H, Hara S, Mizuta K, Sakuma Y, Sata N, Takahashi M. Ferroptosis driven by radical oxidation of n-6 polyunsaturated fatty acids mediates acetaminophen-induced acute liver failure. Cell Death Dis. 2020;11:144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 173]  [Article Influence: 43.3]  [Reference Citation Analysis (0)]
24.  Ji Y, Si W, Zeng J, Huang L, Huang Z, Zhao L, Liu J, Zhu M, Kuang W. Niujiaodihuang Detoxify Decoction inhibits ferroptosis by enhancing glutathione synthesis in acute liver failure models. J Ethnopharmacol. 2021;279:114305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
25.  Wang Y, Chen Q, Shi C, Jiao F, Gong Z. Mechanism of glycyrrhizin on ferroptosis during acute liver failure by inhibiting oxidative stress. Mol Med Rep. 2019;20:4081-4090.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
26.  Siregar AS, Nyiramana MM, Kim EJ, Cho SB, Woo MS, Lee DK, Hong SG, Han J, Kang SS, Kim DR, Choi YJ, Kang D. Oyster-Derived Tyr-Ala (YA) Peptide Prevents Lipopolysaccharide/D-Galactosamine-Induced Acute Liver Failure by Suppressing Inflammatory, Apoptotic, Ferroptotic, and Pyroptotic Signals. Mar Drugs. 2021;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
27.  Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discov. 2020;6:36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 483]  [Article Influence: 120.8]  [Reference Citation Analysis (0)]
28.  Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2019;133:144-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 401]  [Cited by in F6Publishing: 766]  [Article Influence: 153.2]  [Reference Citation Analysis (0)]
29.  Wang K, Sun Q, Zhong X, Zeng M, Zeng H, Shi X, Li Z, Wang Y, Zhao Q, Shao F, Ding J. Structural Mechanism for GSDMD Targeting by Autoprocessed Caspases in Pyroptosis. Cell. 2020;180:941-955.e20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 383]  [Article Influence: 95.8]  [Reference Citation Analysis (0)]
30.  Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, Cao L, Xie M, Ran Q, Kroemer G, Wang H, Billiar TR, Jiang J, Tang D. Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe. 2018;24:97-108.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 382]  [Article Influence: 63.7]  [Reference Citation Analysis (0)]
31.  Liu T, Yang L, Gao H, Zhuo Y, Tu Z, Wang Y, Xun J, Zhang Q, Zhang L, Wang X. 3,4-dihydroxyphenylethyl alcohol glycoside reduces acetaminophen-induced acute liver failure in mice by inhibiting hepatocyte ferroptosis and pyroptosis. PeerJ. 2022;10:e13082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
32.  Borude P, Bhushan B, Chavan H, Weemhoff JL, Jaeschke H, Krishnamurthy P, Apte U. P53 Regulates Progression of Injury and Liver Regeneration After Acetaminophen Overdose. FASEB J. 2017;31.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ. 2022;29:895-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 229]  [Article Influence: 114.5]  [Reference Citation Analysis (0)]
34.  Chen D, Chu B, Yang X, Liu Z, Jin Y, Kon N, Rabadan R, Jiang X, Stockwell BR, Gu W. iPLA2β-mediated lipid detoxification controls p53-driven ferroptosis independent of GPX4. Nat Commun. 2021;12:3644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 170]  [Article Influence: 56.7]  [Reference Citation Analysis (0)]
35.  Wang C, Liu T, Tong Y, Cui R, Qu K, Liu C, Zhang J. Ulinastatin protects against acetaminophen-induced liver injury by alleviating ferroptosis via the SIRT1/NRF2/HO-1 pathway. Am J Transl Res. 2021;13:6031-6042.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Castro RE, Ferreira DM, Afonso MB, Borralho PM, Machado MV, Cortez-Pinto H, Rodrigues CM. miR-34a/SIRT1/p53 is suppressed by ursodeoxycholic acid in the rat liver and activated by disease severity in human non-alcoholic fatty liver disease. J Hepatol. 2013;58:119-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 273]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
37.  Tian XF, Ji FJ, Zang HL, Cao H. Activation of the miR-34a/SIRT1/p53 Signaling Pathway Contributes to the Progress of Liver Fibrosis via Inducing Apoptosis in Hepatocytes but Not in HSCs. PLoS One. 2016;11:e0158657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 65]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
38.  Ma S, Sun L, Wu W, Wu J, Sun Z, Ren J. USP22 Protects Against Myocardial Ischemia-Reperfusion Injury via the SIRT1-p53/SLC7A11-Dependent Inhibition of Ferroptosis-Induced Cardiomyocyte Death. Front Physiol. 2020;11:551318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 114]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
39.  Aktaş HS, Ozel Y, Ahmad S, Pençe HH, Ayaz-Adakul B, Kudas I, Tetik S, Şekerler T, Canbey-Göret C, Kabasakal L, Elcioglu HK. Protective effects of resveratrol on hepatic ischemia reperfusion injury in streptozotocin-induced diabetic rats. Mol Cell Biochem. 2019;460:217-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]